EP2798069B1 - Porcine knob xenotype chimeric adenoviral vector for dendritic cell infection - Google Patents

Porcine knob xenotype chimeric adenoviral vector for dendritic cell infection Download PDF

Info

Publication number
EP2798069B1
EP2798069B1 EP12858372.1A EP12858372A EP2798069B1 EP 2798069 B1 EP2798069 B1 EP 2798069B1 EP 12858372 A EP12858372 A EP 12858372A EP 2798069 B1 EP2798069 B1 EP 2798069B1
Authority
EP
European Patent Office
Prior art keywords
seq
knob
ad5luc1
fiber
immunizing antigen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP12858372.1A
Other languages
German (de)
French (fr)
Other versions
EP2798069A2 (en
EP2798069A4 (en
Inventor
David Curiel
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Washington University in St Louis WUSTL
Original Assignee
Curiel David
University of Washington
Washington University in St Louis WUSTL
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Curiel David, University of Washington, Washington University in St Louis WUSTL filed Critical Curiel David
Publication of EP2798069A2 publication Critical patent/EP2798069A2/en
Publication of EP2798069A4 publication Critical patent/EP2798069A4/en
Application granted granted Critical
Publication of EP2798069B1 publication Critical patent/EP2798069B1/en
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5256Virus expressing foreign proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10322New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/60Vectors comprising as targeting moiety peptide derived from defined protein from viruses
    • C12N2810/6009Vectors comprising as targeting moiety peptide derived from defined protein from viruses dsDNA viruses
    • C12N2810/6018Adenoviridae
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Description

    Related Applications
  • This application claims priority to US Provisional Patent Application 61/576,116 filed 15 December 2011 .
  • Government Support
  • This work was made with the support of Grant 5R33AI076096-06 from the National Institutes of Health. The government of the United States of America may have certain rights in this work.
  • Incorporation-by-reference of Sequence Listing
  • The Sequence Listing, which is a part of the present disclosure, includes a computer readable form and a written sequence listing comprising nucleotide and/or amino acid sequences. The subject matter of the Sequence Listing is incorporated herein by reference in its entirety. The information recorded in computer readable form is identical to the written sequence listing.
  • Introduction
  • Dendritic cells (DCs) are an important component of innate immunity. DCs are potent antigen presenting cells (APCs) with the ability to initiate the primary immune response. Banchereau, J., et al., Annu. Rev. Immunol. 18: 767-811, 2000. In addition to their role in local innate immune responses, DCs play a crucial role in adaptive immune response by priming the immune response or by inducing tolerance. Humans have DCs with different phenotypes that are distributed throughout the body and reside at the site of potential pathogen entry or tissue injury, where they differentiate into mature DCs (Li, K., et al., Mol. Immunol. 48: 1121-1127, 2011). During maturation, DCs undergo phenotypic and functional changes allowing them to increase their antigen presenting and increase their expression of co-stimulatory molecules (Rea, D., et al., J. Immunol. 166: 5236-5244, 2001). Bacterial and viral compounds have been identified as major DC maturation signals (Sallusto, F. and Lanzavecchia, A., J. Exp. Med.182: 389-400, 1994; Hartmann, G., et al., Proc. Natl. Acad. Sci. USA. 96: 9305-9310, 1999; Sparwasser, T., et al., Eur. J. Immunol. 28: 2045-2054, 1998; Verdijk, R.M., et al., J. Immunol. 163: 57-61, 1999; Cella, M., et al., J. Exp. Med. 189: 821-829, 1999).
  • Since DCs have a unique ability to prime an immune response, targeting them in immune intervention strategies against infectious diseases as well as cancer has shown promise (Palucka, K., et al., J. Immunol. 186: 1325-1331, 2011). Multiple approaches have been developed to deliver antigens to DCs for presentation including transfection with DNA or RNA and gene transfer via recombinant vectors (Fong, L., et al., Annu. Rev. Immunol. 18: 245-273, 2000; Gong, J., et al., Nat. Med. 3: 558-561, 1997). Genetic modification of DCs with recombinant viruses offers major advantages including persistent antigen presentation over time and exposure to potentially immune-activating viral components (Sloan, J.M., et al. Cancer Gene Ther. 9: 946-950, 2002). Clinical trials have shown treatment with adenovirus-based vectors are safe and with the development of transductionally targeted, selectively replicating vectors to be increasingly effective against diseases (Paul, C.P.L., et al., Cancer Biol. Ther. 7: 786-793, 2008). Paul et al. showed that replacing the fiber knob of Ad5 with certain non-human knobs enhanced infectivity of human glioma cell populations and primary tumor cells.
  • Rieneke Van De Ven et al., (J. Immunother. 2009; 32(9): 895-906) disclose selective transduction of mature DC in human skin and lymph nodes by CD80 / CD86-targeted fiber-modified Adenovirus-5/3.
  • C.P.L. Paul et al., (Cancer Biology & Therapy 7:5, 786-793; May 2008) characterizes infectivity of knob-modified adenoviral vectors in glioma.
  • Infection of DCs with adenovirus is limited because human DCs lack the native adenovirus receptor, coxsackie-adenovirus receptor (CAR). Ad5 carrying subgroup B Ad fibers are more potent than classical Ad5 for gene transfer and expression in human DCs (Rea, D., et al., J. Immunol. 166: 5236-44, 2001). In order to achieve meaningful therapeutic efficacy of adenovirus-based therapies, new approaches for infection of human DCs are required
  • Summary
  • The present inventors have developed dendritic cells comprising modified adenoviral vectors which can infect dendritic cells with much greater infectivity compared to wild type adenovirus, as defined in the claims. In various configurations, an adenoviral vector of the present teachings can be a chimeric adenovirus which comprises a fiber comprising a tail, a shaft and a knob, wherein the knob is a porcine knob. The tail is an Ad5 tail, and the shaft is an Ad5 shaft, so that a fiber of the present teachings comprises a human Ad5 tail, a human Ad5 shaft, and a porcine knob. In some embodiments a chimeric adenovirus of the present teachings can bind dendritic cells using a receptor other than a CAR receptor. In some embodiments, a chimeric adenovirus of the present teachings can bind dendritic cells using a receptor other than an integrin receptor. In some embodiments, a chimeric adenovirus of the present teachings can bind dendritic cells using a receptor other than either a CAR receptor or an integrin receptor. In some embodiments, a chimeric adenovirus of the present teachings can comprise a fiber comprising a knob, wherein the knob comprises a galectin domain. In some configurations, a galectin domain can bind to one or more carbohydrates, such as a carbohydate comprising lactose and N-acetyl-lactosamine units. In some configurations, a galectin domain comprised by a knob of the present teachings can bind a carbohydrate structure selected from the group consisting of Galß1-4GlcNAcß1-3Galß1-4GlcNAcß1-3Galß1-4GlcNAc [tri(Nacetyl-lactosamine)], GlcNAcα1-4Galß1-4GlcNAcß1-3Galß1-4GlcNAcß1-3Galß1-4GlcNAc, Galß1-4GlcNAcß1-3Galß1-4Glc (lacto-N-neotetraose), Galα1-4Galß1-4GlcNAcß1-3Galß1-4Glc and Galß1-4GlcNAcβ1-3Galß1-3GlcNAc. In some configurations, a chimeric adenovirus comprising a fiber comprising a knob of the present teachings can bind a cell-surface glycoprotein comprising a carbohydrate structure such as, without limitation, Galß1-4GlcNAcß1-3Galß1-4GlcNAcß1-3Galß1-4GlcNAc[tri(Nacetyl-lactosamine)], GlcNAcα1-4Galß1-4GlcNAcß1-3Galß1-4GlcNAcß1-3Galß1-4GlcNAc, Galß1-(lacto-N-neotetraose), Galα1-4Galß1-4GlcNAcß1-3Galß1-4Glc, Galß1-4GlcNAcß1-3Galß1-3GlcNAc or a combination thereof.
  • In various embodiments, a galectin domain comprised by a knob of the present teachings can bind to Lacto-N-neotetraose with a dissociation constant of 193±9µM, 3-aminopropyl-lacto-N-neotetraose with a dissociation constant of 303±4µM, 2-azidoethyl-di(N-acetyl-lactosamine) with a dissociation constant of 309±9µM, or 2-aminoethyl-tri(N-acetyl-lactosamine) with a dissociation constant of 308±40µM.
  • In various aspects, cellular uptake by a dendritic cell of DNA of a chimeric Ad5 of the present teachings can be greater than that of a wild-type Ad5. In various aspects, cellular uptake by a dendritic cell of DNA of a chimeric Ad5 of the present teachings can be as great, or greater than, that of an adenovirus comprising a knob comprising an RGD sequence. In various aspects, cellular uptake by a dendritic cell of DNA of a chimeric Ad5 can be as great, or greater than, that of an adenovirus comprising a knob comprising an adenovirus comprising a type 35 fiber.
  • In some embodiments, a dendritic cell of the present teachings can comprise a chimeric adenovirus-5 (Ad5) viral genome, wherein the chimeric Ad5 genome encodes a) a fiber comprising a tail, a shaft and a knob, wherein the knob is a porcine knob; and b) a promoter operably linked to a heterologous sequence encoding an antigen peptide.
  • In some embodiments, the present teachings include ex vivo cell cultures comprising a dendritic cell, in particular a human dendritic cell, wherein the dendritic cell comprises nucleic acid sequences encoding Ad5sequences encoding a modified tail that includes a porcine knob sequence, as described herein .
  • In some embodiments, the present teachings include a vaccine comprising an Ad5 modified to comprise a porcine knob, as well as an antigen peptide sequence such as, for example, an antigen peptide consisting of a linear peptide of from at least 8 up to 15 amino acids, for example 9 amino acids such as a peptide sequence set forth in table 1.
  • In some embodiments, a vaccine of the present teachings can comprise a dendritic cell comprising Ad5 sequences encoding a modified tail protein, such as a tail protein comprising a porcine knob. In some configurations, a vaccine can comprise dendritic cells autologous to a subject such as a human subject, in which dendritic cells obtained from the subject can be grown and/or infected with a modified Ad5 of the present teachings in a cell culture ex vivo. Such cells can be administered to a subject, such as, for example, the donor of the dendritic cells, using methods well known to skilled artisans.
  • In some embodiments of the present teachings, a subject such as a human can receive a vaccination through administration of a modified Ad5 of the present teachings. In some embodiments, a subject such as a human can receive a vaccination through administration of dendritic cells infected with an Ad5 of the present teachings. In some configurations, the dendritic cells can be autologous dendritic cells.
  • In various configurations, an antigen peptide sequence can comprise or consist of from about 8, at least 8 up to 15, or about 15, contiguous amino acids. In some configurations, an antigen peptide sequence can comprise or consist of 9 contiguous amino acids. In various aspects, a peptide sequence can be that of a protein fragment, wherein the protein is a pathogen protein or a cellular protein, such as, for example, a protein expressed by a cancer cell. In some aspects, an antigen can comprise an antigen peptide such as that of an HLA-A restricted peptide or HLA-B restricted peptide. In some aspects, an antigen peptide can comprise or consist of a sequence as set forth in Table 1. Table 1: Antigen Peptide Sequences
    Name Source Sequence Identification
    CMVpp65 Cytomegalovirus NLVPMVATV SEQ ID NO: 1
    EBV BMLF I Ebstein-Barr virus GLCTLVAML SEQ ID NO: 2
    fluM1 Influenza A virus GILGFVFTL SEQ ID NO: 3
    G209-2M human melanoma IMDQVPFSV SEQ ID NO: 4
    G280-9V human melanoma YLEPGPVTV SEQ ID NO: 5
  • Various embodiments of the present teachings include a dendritic cell comprising a chimeric adenovirus-5 (Ad5) viral genome, wherein said chimeric Ad5 genome encodes a) a fiber comprising a tail, a shaft and a knob, wherein the knob is a porcine knob; and b) a promoter operably linked to a heterologous sequence encoding an immunizing antigen. As used herein, an "immunizing antigen" is a protein, polypeptide or oligopeptide that can stimulate an immune response in a body such as a human body.
  • In some embodiments, the present teachings include an ex vivo cell culture comprising a dendritic cell comprising a chimeric Ad5 genome which encodes a) a fiber comprising a tail, a shaft and a knob, wherein the knob is a porcine knob; and b) a promoter operably linked to a heterologous sequence encoding an immunizing antigen. For example, an antigen that can bind a corresponding MHC class I heavy chain or MHC class I-like antigen presenting molecule such as CD1 (Altamirano, M.M., et al., Proc. Nat'1 Acad. Sci. 98: 3288-3293, 2001). In some aspects, an immunizing antigen can be that of a peptide which can be presented by an MHC class I molecule.
  • In some embodiments, the present teachings include vaccines, wherein a vaccine comprises a dendritic cell comprising a chimeric Ad5 genome which encodes a) a fiber comprising a tail, a shaft and a knob, wherein the knob is a porcine knob; and b) a promoter operably linked to a heterologous sequence encoding an immunizing antigen. In various configurations, an immunizing antigen can be a peptide comprising or consisting of about 8, from 8 to 15, or about 15 contiguous amino acids. In various configurations, an immunizing antigen can be a peptide comprising or consisting of 9 contiguous amino acids, or about 9 contiguous amino acids. In various configurations, an immunizing antigen can be a peptide comprising or consisting of a sequence selected from the group consisting of NLVPMVATV (SEQ ID NO: 1), GLCTLVAML (SEQ ID NO: 2), GILGFVFTL (SEQ ID NO: 3), IMDQVPFSV (SEQ ID NO: 4) and YLEPGPVTV (SEQ ID NO: 5).
  • Various embodiments of the present teachings include a chimeric Ad5 comprising: a) a fiber comprising a tail, a shaft and a knob, wherein the knob is a porcine knob; and b) a promoter operably linked to a heterologous sequence encoding an immunizing antigen. In various configurations, an immunizing antigen can comprise or consist of a sequence of a protein expressed by a cell at a level associated with a disease. In various configurations, an immunizing antigen can comprise or consist of a sequence of a protein expressed by a cancer cell at a level associated with a cancerous phenotype. In various configurations, an immunizing antigen can comprise or consist of about 8, from 8 to 15, or about 15 contiguous amino acids. In various configurations, an immunizing antigen can comprise or consist of 9, or about 9 contiguous amino acids. In various configurations, an immunizing antigen can comprise or consist of a peptide having a sequence selected from the group consisting of NLVPMVATV (SEQ ID NO: 1), GLCTLVAML (SEQ ID NO: 2), GILGFVFTL (SEQ ID NO: 3), IMDQVPFSV (SEQ ID NO: 4) and YLEPGPVTV (SEQ ID NO: 5).
  • Brief description of the Drawings
    • FIG. 1 illustrates the design of the Ad5Luc1-PK chimeric fiber.
    • FIG. 2 illustrates molecular validation of Ad5Luc1-PK virions.
    • FIG. 3 illustrates fluorescence micrographs of CAR-negative CHO and CAR-positive CHO-hCAR cell lines.
    • FIG. 4 illustrates that gene transfer of Ads-PK vectors is CAR-independent.
    • FIG. 5 illustrates that Ad5Luc1-PK uses carbohydrate binding domains for gene transfer.
    • FIG. 6 illustrates that Ad5Luc1-PK-mediated gene delivery is mediated by glycans containing lactose.
    • FIG. 7 illustrates Ad5Luc1-PK infectivity in murine dendritic cells.
    • FIG. 8 illustrates Ad5Luc1-PK infectivity in Cynomolgus macaque dendritic cells.
    • FIG. 9 illustrates Ad5Luc1-PK infectivity in human dendritic cells.
    • FIG. 10 illustrates Ad5GFP-PK infectivity in human dendritic cells.
    Detailed Description
  • The present inventors disclose a chimeric adenovirus and methods of transforming dendritic cells therewith. These methods, in various configurations, can enhance infectivity adenovirus towards human dendritic cells. In various embodiments, a porcine knob, which contains a galectin domain, is able to bind to carbohydrate moieties on the cell surface of dendritic cells. In some configurations, the carbohydrate moieties can comprise lactose and N-acetyl-lactosamine units. Furthermore, in some configurations the lactose and N-acetyl-lactosamine units can be Galß1-4GlcNAcß1-3Galß1-4GlcNAcß1-3Galß1-4GlcNAc [tri(Nacetyl-lactosamine)], GlcNAcα1-4Galß1-4GlcNAcß1-3Galß1-4GlcNAcß1-3Galß1-4GlcNAc, Galß1-4GlcNAcß1-3Galß1-4Glc (lacto-N-neotetraose), Galα1-4Galß1-4GlcNAcß1-3Galß1-4Glc, Galß1-4GlcNAcß1-3Galß1-3GlcNAc or a combination thereof. In various configurations the galectin domain can bind to Lacto-N-neotetraose with a dissociation constant of 193+9µM, to 3-aminopropyl-lacto-N-neotetraose with a dissociation constant of 303±4µM, to 2-azidoethyl-di(N-acetyl-lactosamine) with a dissociation constant of 309±9µM, or to 2-aminoethyl-tri(N-acetyl-lactosamine) with a dissociation constant of 308±40µM (the SPR response in µRIU).
  • Monocytes and dendritic cells (DCs), such as freshly isolated human blood myeloid DCs, plasmacytoid DCs and monocyte-derived DCs lack CAR expression, but Langerhans cells and dermal DCs from skin express CAR. Furthermore, monocyte-derived DCs have lower CD46 expression then dermal DCs, Langerhans DCs, myeloid DCs, and plamacytoid DCs. Expression of CAR and CD46 (the subgroup C and B adenovirus receptors) on dendritic cell surfaces can be measured using FACS in cell lines. The correlation between infectivity enhancement and expression levels of CAR and CD46 can be determined.
  • For example, infectivity of a panel of fiber-modified Ads that are CAR-independent can be compared in a variety of cancer cell types. The fiber-modified Ads can be examined to determine gene transfer to dendritic cell lines and compared with a tropism-modified Ad vector, Ad5/3, which encodes a fiber composed of the native Ad5 tail and shaft domains, but the fiber knob domain from Ad3.
  • In some configurations, Ad5Luc1-PK and Ad5Luc1-CK1 fiber-modified adenovirus vectors of the present teachings can be Ad vectors with enhanced infectivity toward dendritic cells in comparison to an Ad5 comprising a wild-type knob. For example, three of the fiber-modified vectors, Ad5Luc1-PK, Ad5Luc1-CK1 and Ad5/3, can exhibit enhanced infectivity towards human dendritic cells compared to Ad5Luc1. In some configurations, Ad5Luc1-PK and Ad5Luc1-CK1 can have more than a 10-fold greater infectivity compared to that of Ad5/3. In some configurations, an Ad5Luc1-PK can have a greater infectivity compared to that of an Ad5 with a type 35 fiber described in Rea et al. (J. Immunol. 166: 5236-44, 2001).
  • Methods
  • Methods and compositions described herein utilize laboratory techniques well known to skilled artisans. Such techniques can be found in laboratory manuals such as Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 3rd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2001; Spector, D. L. et al., Cells: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1998; Harlow, E., Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1999.
  • The following materials and methods are used in some experiments reported herein.
  • Plasmid construction. A 1,750-bp region containing the PAdV-4 fiber knob and carbohydrate binding domains (amino acids 121-703) of the fiber protein was amplified from cell lysates containing wild type PAdV-4 virus obtained from the US Department of Agriculture National Veterinary Services Laboratory (Ames, Iowa) using the following primers: (PAd4 knob fwd) 5'-TGTGGACGGGGCCTGCTC-3'(SEQ ID: 6) and (PAd4 knob rev) 5'-TTTATTACAGTATCTGAGG-3'(SEQ ID: 7). Plasmid pSHAFT, a cloning vector containing the Ad5 fiber gene with the knob region deleted and replaced by a small linker containing Sma1 and EcoICR1 restriction sites (Krasnykh, V.N., et al., J. Virol. 70: 6839-46, 1996), was linearized by Sma1 and EcoICR1 digestion, leaving two blunt ends. Following gel purification, the PAdV-4 knob domain PCR product was ligated into linearized pSHAFT resulting in pSHAFT-PK and positive clones were screened for correct orientation via restriction enzyme digest. This plasmid contains the chimeric fiber gene encoding the complete Ad5 fiber shaft in-frame with the PAdV-4 knob domain. A stop codon and polyadenylation sequence is present at the 3' end. The chimeric fiber gene in pSHAFT was digested with Ncol and MunI to liberate the DNA fragment containing the carboxy terminus of the HAdV-5 shaft and the PAdV-4 knob domain. This fragment was ligated into the NcoI-MunI-digested fiber shuttle vector pNEB.PK.3.6 (Krasnykh, V.N., et al., J. Virol. 70: 6839-46, 1996), resulting in pNEB.PK.3.6-PK.
  • Generation of recombinant adenovirus. The recombinant Ad5Luc1-PK genome containing the chimeric PAdV-4 fiber gene was derived by homologous recombination in E. coli BJ5183 with SwaI-linearized rescue plasmid pVK700 (Belousova, N., et al,. J. Virol. 76: 8621-31, 2002) and the fiber-containing PacI-KpnI-fragment of pNEB.PK.3.6-PK, essentially as described (Krasnykh, V., et al., J. Virol. 72: 1844-52, 1998). Plasmid pVK700 is derived from pTG3602 (Chartier, C., et al., J. Virol. 70: 4805-10, 1996), but contains an almost complete deletion of the fiber gene and contains a firefly luciferase reporter gene driven by the cytomegalovirus immediate early promoter in place of the E1 region. The recombinant genome of Ad5GFP1-PK containing the chimeric PAdV-4 fiber gene was derived by homologous recombination in E. coli BJ5183 with fiber shuttle plasmid pKan3.1-PK which contains the same chimeric fiber gene as pNEB.PK.3.6-PK described above, and SwaI-linearized rescue plasmid pVK900 (Murakami, M., et al., Virol. 407: 196-205, 2010). Plasmid pVK900 is a fiber-deleted HAdV-5 genome plasmid essentially the same as pVK700 except that EGFP is encoded in the E1 region (supplied by Victor Krasnykh, University of Texas MD Anderson Cancer Center). All genomic clones were sequenced and analyzed by PCR prior to transfection of HEK 293 cells. Ad5Luc1 is a replication defective E1-deleted Ad vector containing a firefly luciferase reporter gene driven by a cytomegalovirus promoter (Krasnykh, V., et al., J. Virol. 75: 4176-83, 2001). All vectors were propagated on HEK 293 cells and purified by equilibrium centrifugation in CsCl gradients by standard protocols. Viral particle concentration was determined at 260 nm by the method of Maizel et al. (Maizel, J.V., et al., Virol. 36: 115-25, 1968) by using a conversion factor of 1.1x1012 viral particles/absorbance unit.
  • Generation of monocyte-derived DC.
  • DC medium is RPMI + 2mM glutamine + HEPES + non-essential amino-acids + Pen/Strep + 1% AB sera.
  • Peptide pulsing medium is Stemline + 2mM L-glutamine + Pen/Strep + 1% AB sera A-DC generation.
    1. 1) Fresh PBMC are isolated from blood, buffy coats or leukopheresis as instructed on protocol. Frozen PBMC (yield ∼ 2×108 cells/vial) are thawed, counted. Fresh or frozen PBMC are adjusted to 5x106/ml in DC media. Transfer 40ml (2x108 cells) to a T175 flask and incubate for 2h at 37°C.
    2. 2) Remove non-adherent cells and transfer to a 50ml conical tube. Wash T175 gently 2X with 25 ml PBS and transfer to another 50 ml conical tube.
      1. a. These PBL can be discarded or can be used as a source of T cells (see Primary CTL stimulation protocol).
    3. 3) To T175 ml flask add 30 ml DC media containing 100ng/ml GM-CSF (Leukine) and 20ng/ml IL-4 (CellGenix). Incubate cells at 37°C 5% CO2.
    4. 4) On day 3 feed cells with 10ml DC medium containing 100ng/ml GM-CSF and 20ng/ml IL-4.
    5. 5) Harvest cells on day 6 by gently rocking flask back and forward; collect non-adherent and loosely adherent cells and transfer to a 50ml conical tube. Wash T175 flask gently 2X with 25ml PBS and transfer to another 50 ml conical tube. Spin at 1500 RPM for 5 min, aspirate supernatant, resuspend cells in DC medium (1-2 ml/tube), pool cells and count. DC are adjusted to 2x106/ml in DC media containing 200ng/ml GM-CSF and 40ng/ml IL-4 (2X concentration)
      Yield: frozen PBMC ∼2-5 x 106 DC/flask; from fresh cells ∼107 DC/flask.
      B-CD40L/IFN-γ maturation
    6. 6) Immature DC consist of cells grown in GM-CSF and IL-4; diluted 1:1 vol with DC media.
    7. 7) J558-muCD40L or K562-huCD40L are used for maturation. Cells are irradiated (5,000 RADS for J558 or 10,000RADS for K562), spun and resuspend in DC media at a 4x105 cells/ml in DC media.
    8. 8) Mix 1:1 vol of DC to CD40L-expressing cells, up to 4ml per well of a 6 well tray (Ultra-low #3471). Ratio of DC to CD40L cells is 5DC:1CD40L-expressing cell.
    9. 9) Add 100u/ml IFN-g (Actimmune). Incubate for 24-48h
    10. 10) Harvest DC, save undiluted supernatant for assessment of cytokine production. Wash cells once, and resuspend in Peptide pulsing media if cells are to be use in stimulation of T cells.
    NOTE: Immature (GM-CSG/IL-4) and mature (GM-CSG/IL-4 + CD40L+ IFN-γ) are characterized by production of IL-12p70 (ELISA) and up-regulation of HLA-DR, CD86 and CD83 (all markers vs. CD11c, by FACS).
  • PCR Analysis of the Fiber Region. Genomic DNA contained in Ad5Luc1, Ad5Luc1-PK and PAdV-4 viral particles was used as templates for PCR amplification of fiber genes using a HAdV-5-specific primer set: (fwd) 5'-CAGCTCCATCTCCTAACTGT-3'(SEQ ID: 8) and (rev) 5'-TTCTTGGGCAATGTATGAAA-3'(SEQ ID: 9) and a PAdV-4-specific primer set: (fwd) 5'-TGTGGACGGGGCCTGCTC-3' (SEQ ID: 10) and (rev) 5'-TTTATTACAGTATCTGAGG-3'(SEQ ID: 11). Wild type PAdV-4 virus was used as a positive control.
  • Western Blot Analysis. Purified Ad virions (5.0 x 109) were diluted in Laemmli buffer and incubated at room temperature (unboiled samples) or 95°C (boiled samples) for 10 minutes and loaded onto a 4-20% gradient SDS-polyacrylamide gel (Bio-Rad, Hercules, CA). Following electrophoretic separation, Ad capsid proteins were electroblotted onto a PVDF membrane and detected with a 4D2 monoclonal anti-fiber tail primary antibody diluted 1/3000 (Lab Vision, Freemont CA). Immunoblots were developed by addition of a secondary horseradish peroxidase-conjugated anti-mouse immunoglobulin antibody at a 1/3000 dilution (Dako Corporation, Carpentaria, CA), followed by incubation with 3-3'-diaminobenzene peroxidase substrate, DAB, (Sigma Chemical Company, St. Louis, MO).
  • Ad-Mediated Gene Transfer Assays. Cells were plated in 24-well plates and were transduced for 1 hour at 37°C with each Ad vector diluted to 100-300 viral particles/cell in 500 µL of transduction media containing 2% FBS. Following the incubation, virus-containing media was replaced with fresh media containing 2% FBS and cells were maintained at 37°C in an atmosphere containing 5% CO2. Cells were harvested 24 hours post-transduction in passive lysis buffer and gene transfer was determined using a luciferase activity assay system (Promega, Madison, WI). Fluorescence microscopy was performed with an inverted IX-70 microscope (Olympus) using a 20X objective. Cells were infected with Ad5GFP1-PK for 24 hours prior to imaging.
  • For experiments assessing the competitive inhibition containing of vector binding to cells, recombinant fiber knob protein (Glasgow, J.N., et al., Virol. 324: 103-16, 2004) at 0.5, 5.0 and 50 µg/ml final concentration or recombinant PAdV-4 carbohydrate binding domain (CBD) protein [20] at 0.5, 5.0, 50 and 100 µg/ml was incubated with various cell lines at 37°C in media containing 2% FBS for 20 minutes prior to the addition of HAdV-5 vectors. Following transduction with Ad vectors, cells were rinsed with media to remove unbound virus and blocking proteins, and were maintained at 37°C in an atmosphere containing 5% CO2.
  • To inhibit glycosylation of cellular proteins, chemical inhibitors of glycosylation were used. CHO-Pro5 cells were incubated with medium containing 10 µg/ml swainsonine (Sigma, Saint Louis, MO., S8195) and/or 1 µg/ml benzyl-α-GalNAc (Sigma B4894) for 24 hr at 37 °C, followed by addition of Ad vectors in media containing 2% FBS.
  • Examples
  • The present teachings including descriptions provided in the Examples that are not intended to limit the scope of any claim or aspect. Unless specifically presented in the past tense, an example can be a prophetic or an actual example. The following non-limiting examples are provided to further illustrate the present teachings. Those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments that are disclosed and still obtain a like or similar result without departing from the scope of the present teachings.
  • Example 1
  • This example illustrates the generation of a fiber-modified HAdV-5 vector containing the PAdV-4 knob and carbohydrate binding domains.
  • The fiber protein of PAdV-4 NADC-1 is comprised of a homotrimer of 703 amino acids (FIG. 1). Predicted functional domains include a tail domain (residues 1-37) containing a penton interaction sequence, a short shaft domain (residues 38-120) with six predicted triple beta-spiral repeats (van Raaij, M.J., et al. Virol. 262: 333-43, 1999) and a fiber knob domain homologous to other Ad fiber knob domains (residues 121-287) (Guardado-Calvo, P., et al. Acta. Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 65: 1149-52, 2009). This fiber also contains a unique C-terminal domain composed of two tandem carbohydrate binding domains (CBDs) (residues 393-681) that bind carbohydrates containing lactose and N-acetyl-lactosamine units (Guardado-Calvo, P., et al. J. Virol. 84: 10558-68, 2010). Almost all mastadenoviruses contain a conserved threonine-leucine-tryptophan-threonine (TLWT) (SEQ ID NO: 12) motif at the N-terminus of the fiber knob domain, and in human Ad2 and Ad5 a flexible region separating the shaft and the knob domains precedes this motif (van Raaij, M.J., et al. Nature. 401: 935-8, 1999). The modular fiber structure was used to substitute the coding region of the PAdV-4 knob and CBD domains for the HAdV-5 fiber knob sequence while retaining the TLWT motif common to both fibers (Renaut, L., et al. Virol. 321: 189-204, 2004). A recombinant E1-deleted HAdV-5 genome (Ad5Luc1-PK) containing the chimeric HAdV-5 shaft/PAdV-4 fiber gene and a firefly luciferase reporter gene controlled by the CMV immediate early promoter/enhancer were constructed. The Ad5Luc1-PK vector was rescued following transfection of HEK 293 cells and large-scale preparations of Ad5Luc1-PK and the Ad5Luc1 control vector were produced and purified by double CsCl gradient centrifugation. Ad5Luc1-PK viral particle concentration in full preparations ranged from 1.2 x 1011 to 1.25 x 1012 viral particles/ml, similar to that of the Ad5Luc1 control vector containing the HAdV-5 wild type fiber. The Ad5Luc1 vector is isogenic to Ad5Luc1-PK except for the fiber protein.
  • The fiber genotypes of Ad5Luc1 and Ad5Luc1-PK vectors were confirmed via diagnostic PCR using primer pairs specific for the fiber knob domain and genomes from purified virions as PCR templates. Genomic DNA from wild type PAdV-4 was used as a positive control. The expected PCR products were observed for the wild type HAdV-5 fiber knob domain (530 bp) and the PAdV-4 fiber knob and CBD domains (1,750 bp) (FIG. 2A).
  • SDS-PAGE was performed followed by western blot analysis on purified viral particles to verify that Ad5Luc1-PK virions contain correctly trimerized chimeric fiber proteins (FIG. 2B). Blots were probed with a monoclonal primary antibody (4D2) directed against the fiber tail domain common to both HAdV-5 and chimeric fiber molecules. In samples that were not heat denatured (FIG. 2B, unboiled) bands were observed at 183 kDa and an estimated 250 kDa, corresponding to trimers of the HAdV-5 fiber and chimeric fibers, respectively. Further, bands in boiled samples resolved at apparent molecular masses of 60 kDa for the wild type HAdV-5 fiber and 90 kDa for the chimeric fiber in Ad5Luc1-PK, representing fiber monomers.
  • Example 2
  • This example illustrates that Ad5-PK vector infectivity is independent of CAR.
  • High resolution crystal structure analysis has shown that the A-B loop in the N-terminal region of the PAdV-4 knob domain is structurally similar to the A-B loop in the CAR-binding domain in adenoviruses that use CAR as a primary receptor, but that only one CAR-binding residue is conserved (Guardado-Calvo, P., et al. J. Virol. 84: 10558-68, 2010). To investigate whether Ad5-PK vectors exhibit CAR-dependent tropism, gene transfer assays were performed in two cell lines with markedly different levels of CAR expression: CAR-deficient Chinese hamster ovary (CHO) cells and a CHO-derived cell line, CHO-hCAR, which stably expresses human CAR (Bergelson, J.M., et al. Science. 275: 1320-3, 1997). CHO and CHO-hCAR cells were infected with the Ad5GFP control vector and Ad5GFP1-PK, a vector isogenic to Ad5Luc1-PK except that the firefly luciferase reporter gene was replaced with green fluorescent protein, GFP. Fluorescence microscopy showed GFP expression in CHO-hCAR cells but not in CAR-deficient CHO cells (FIG. 3), consistent with native HAdV-5 tropism. In contrast, Ad5Luc1-PK-mediated GFP gene delivery does not depend on CAR expression, as similar number of GFP-positive cells were observed in both the CHO and CHO-hCAR cell lines. Similar gene transfer assays were performed using luciferase-expressing Ad5Luc1 and Ad5Luc1-PK vectors to quantify differences in gene delivery based solely on CAR expression. Ad5Luc1 exhibited the expected CAR-dependent tropism as demonstrated by an 80-fold increase in luciferase activity in CHO-hCAR cells versus CHO cells (FIG. 4A). In contrast, Ad5Luc1-PK provided robust gene delivery to both cell lines. In addition, competitive inhibition of CAR binding with recombinant HAdV-5 knob proteins (50 µg/ml) inhibited over 96% of Ad5Luc1 gene transfer to CHO-hCAR cells, but did not inhibit the gene transfer of Ad5Luc1-PK (FIG. 4B).
  • Example 3
  • This example illustrates that Ad5Luc1-PK uses carbohydrate binding domains for gene transfer.
  • To determine whether the CBDs in the chimeric fiber of Ad5Luc1-PK participate in cellular attachment, competitive inhibition assays were performed using a recombinant protein consisting of the tandem PAdV-4 CBDs (residues 393-703 of the PAdV-4 fiber protein) or recombinant HAdV-5 fiber knob protein as a negative control. Addition of PAdV-4 CBD protein during infection caused a dose-dependent inhibition of Ad5Luc1-PK-mediated gene transfer with a maximum inhibition of 35% at 100 µg/ml (FIG. 5), indicating that the CBDs in the chimeric fiber are responsible for cellular attachment during infection.
  • The CBDs within the PAdV-4 fiber protein bind to lactose, N-actyl-lactosamine and poly-N-acetyl-lactosamine in order of increasing affinity (Guardado-Calvo, P., et al., J. Virol. 84: 10558-68, 2010). However, whether the CBDs in the PAdV-4 chimeric fiber recognize these glycans and use them as a means for viral transduction is not known. Gene transfer assays were performed in the CHO-Lec8 cells which contains mutations in the UDP-galactose transporter/translocase (UGT) gene (Deutscher, S.L., et al., J. Biol. Chem. 261: 96-100, 1986; Stanley, P., Mol. Cell. Biol. 5: 923-29, 1985; Oelmann, S., et al., J. Biol. Chem. 276: 26291-300, 2001), and lack the ability to galactosylate glycoproteins and therefore produce glycoproteins with truncated carbohydrate chains that lack lactose, N-acetyl-lactosamine and poly-N-acetyl-lactosamine. The level of Ad5Luc1 gene delivery was unchanged between CHO-Lec8 cells and the control CHO-Pro5 cells that exhibits normal glycosylation (Fig 6A). In contrast, Ad5Luc1-PK gene delivery to CHO-Lec8 cells was reduced by 64% compared to the control CHO-Pro5 cells, showing that the presence of lactose-containing glycans at the cell surface is critical for Ad5Luc1-PK infectivity.
  • There are two major types of carbohydrate chains on glycoproteins: N-linked glycans linked to asparagine residues and O-linked glycans linked to serine or threonine (Li, H., and d'Anjou, M., Curr. Opin. Biotechnol. 20: 678-84, 2009; Schwarz, F., and Aebi, M., Curr. Opin. Struct. Biol. 21: 576-82, 2011; Dwek, R.A., Chem. Rev. 96: 683-720, 1996). To further investigate the nature of the glycans recognized during Ad5Luc1-PK infection, gene transfer assays were performed following incubation of CHO-Pro5 cells with inhibitors of N-linked glycan synthesis (swainsonine, 10 µg/ml), or O-linked glycan synthesis (benzyl-α-GalNAc, 1 µg/ml) (Elbein, A.D., et al., Proc. Natl. Acad. Sci. USA. 78: 7393-7, 1989; Kuan, S.F., et al., J. Biol. Chem. 264: 19271-7, 1989). The addition of these inhibitors singly or in combination to CHO-Pro5 cells did not alter levels of Ad5Luc1 gene transfer (Fig. 6B). In contrast, Ad5Luc1-PK gene transfer was blocked 35% by benzyl-α-GalNAc pre-treatment, with a minimal (<10%) reduction by swainsonine. Similar results were also observed in A549 cells pre-treated with these inhibitors, suggesting that O-linked cell-surface glycans may be preferred by Ad5Luc1-PK for infection. Collectively, these data show that the CBDs in the chimeric fiber protein of Ad5Luc1-PK directly participate in cellular attachment and that infection is highly dependent on the presence of lactose and/or N-acetyl-lactosamine-containing glycans, consistent with a novel, glycan-mediated cell entry pathway.
  • Example 4
  • This example illustrates enhanced infectivity of Ad5Luc1-PK in murine dendritic cells.
  • To analyze infectivity of Ad5Luc1-PK, gene transfer assays were performed in immature (iDC) and mature (LPS-treated) (mDC) murine dendritic cells. In these experiments, iDC and mDC were infected with the Ad5Luc1 and Ad5Luc1-PK. Ad5Luc1-PK demonstrated a 6-fold increase in luciferase activity in iDC compared to Ad5Luc1, while in mDC Ad5Luc1-PK demonstrated a 8.5 fold increase in luciferase activity as compared to Ad5Luc1 (FIG. 7).
  • Example 5
  • This example illustrates enhanced infectivity of Ad5Luc1-PK in Cynomolgus macaque dendritic cells.
  • To analyze infectivity of Ad5Luc1-PK in Cynomolgus macaque dendritic cells, gene transfer assays were performed in C. macaque dendritic cells. In these experiments, C. macaque dendritic cells were infected with Ad5Luc1, Ad5/3 and Ad5Luc1-PK. Ad5Luc1-PK demonstrated an increase in luciferase activity compared to Ad5/3 and Ad5Luc1 (FIG. 8). Ad5Luc1 (FIG. 7).
  • Example 6
  • This example illustrates enhanced infectivity of Ad5Luc1-PK in human dendritic cells.
  • To analyze infectivity of Ad5Luc1-PK in human dendritic cells, gene transfer assays were performed in immature (iDC) and mature (mDC) human dendritic cells. In these experiments, human dendritic cells were infected with Ad5Luc1, Ad5/3 and Ad5Luc1-PK. In iDC, Ad5/3 demonstrated a 6-fold increase in luciferase activity as compared to Ad5Luc1, while in mDC, Ad5/3 demonstrated a 30-fold increase in luciferase activity as compared to Ad5Luc1 (FIG. 9). In iDC, Ad5Luc1-PK demonstrated an 8-fold increase in luciferase activity compared to Ad5Luc1, while in mDC, Ad5Luc1-PK demonstrated a 40-fold increase in luciferase activity compared to Ad5Luc1 (FIG. 9).
  • Example 7
  • This example illustrates FACS analysis of enhanced infectivity of Ad5GFP1-PK in immature human dendritic cells.
  • To analyze infectivity of Ad5GFP1-PK, gene transfer assays were performed in human dendritic cells. In these experiments, immature human dendritic cells were infected with the Ad5GFP control vector (FIG. 10B, FIG. 10E) and Ad5GFP1-PK (FIG. 10C, FIG 10F), a vector isogenic to Ad5Luc1-PK except that the firefly luciferase reporter gene was replaced with green fluorescent protein, GFP. Uninfected immature human dendritic cells are shown in FIG. 10A and FIG. 10D. Flow cytometry was performed using standard procedures at 24 (FIG. 10 A, B, C) and 48 (FIG. 10 D, E, F) hours after infection. After 24 hours of infection, Ad5GFP infected about 10% of the dendritic cell population (FIG. 10B) as compared to control (FIG. 10A), while Ad5GFP-PK infected 56.5% of the dendritic cell population (FIG. 10C) as compared to control (FIG. 10A). After 48 hours of infection, Ad5GFP infected about 20% of the dendritic cell population (FIG. 10E) as compared to control (FIG. 10D), while Ad5GFP-PK infected 67.7% of the dendritic cell population (FIG. 10F) as compared to control (FIG. 10D).
  • Example 8
  • This example illustrates that xeno-knob-modified adenovirus can have enhanced infectivity in human dendritic cells compared to other adenoviruses.
  • To analyze the efficiency of gene delivery of xenotype-modified adenoviral vectors on dendritic cell lines in vitro, five xeno-knob fiber-modified vectors can be used. In vectors Ad5Luc1-MK, Ad5Luc1-PK, Ad5Luc1-CK1 and Ad5Luc1-CK2 the native Ad5 fiber knob domain can be replaced by the corresponding fiber knob domain from mouse adenovirus type 1, porcine adenovirus, or canine adenovirus type 1 or 2, respectively. In the Ad5Luc1-OvF, the entire Ad5 fiber can be replaced with the fiber from ovine adenovirus 7. A control vector, Ad5Luc1 comprising the native Ad5 fiber, and a fiber-modified vector, Ad5/3, comprising a human Ad serotype 3 fiber knob domain can be used in this example. Both of these vectors can serve as controls to which other fiber-modified Ad vectors can be compared. All Ad vectors can be isogenic except for the fiber gene. The luciferase transgene activity of fiber-modified vectors can be compared to the Ad5Luc1 control vector in dendritic cell lines; plasmacytoid DCs, monocyte-derived DCs, migratory DCs, and lymphoid DCs. Luciferase expression levels can be shown as relative light units (RLU) normalized to that of Ad5Luc1 (Ad5Luc1=100%). Three of the vectors, Ad5Luc1-MK, Ad5Luc1-OvF, and Ad5Luc1-CK2 can augment gene delivery less than 2.5-fold above Ad5Luc1 in dendritic cell lines. In various dendritic cells, the Ad5Luc1-CK1 vector can provide an increase in luciferase transgene activity of at least 7-fold compared to an Ad5Luc1 control. In various dendritic cells, the Ad5Luc1-CK1 vector can provide an increase in luciferase transgene activity of up to 50-fold compared to an Ad5Luc1 control. The Ad5Luc1-PK vector can have an increase in gene delivery from 10-fold up to 20-fold above Ad5Luc1 in the dendritic cell lines. An Ad5/3 vector can enhance gene delivery 14-fold up to 50-fold in dendritic cell lines compared to an Ad5Luc1. Infectivity levels of these two viral vectors (comprising the porcine knob or the canine knob) in the four dendritic cell lines can be similar to or can exceed that of Ad5/3, which had previously been shown to act as a successful fiber-modified vector for enhancement of viral infectivity in human dendritic cells.
  • Example 9
  • This example demonstrates that xeno-knob modified infectivity can be independent of adenovirus native receptor, coxsackie-adenovirus-receptor (CAR).
  • To analyze the infectivity independent of CAR on dendritic cell lines in vitro, Ad5Luc1-PK and Ad5Luc1 can be used, where the Ad5Luc1 can be used as a control. The luciferase transgene activity of Ad5Luc1-PK vector can be compared to the Ad5Luc1 control vector in two dendritic cell lines, monocyte-derived DCs and monocyte-derived DCs that constitutively express high levels of CAR. The luciferase expression levels can be shown as relative light units (RLU). After infection of Ad5Luc, into two dendritic cell lines, the luciferase transgene activity can increase up to 60 fold in monocyte-derived DCs that constitutively express CAR when compared to monocyte-derived dendritic cells. After infection of Ad5Luc1-PK, into monocyte-derived DCs, the luciferase transgene activity can be more then 60-fold increase when compared to Ad5Luc luciferase transgene activity in monocyte-derived DCs. Furthermore, no increase in luciferase transgene activity could (would) be observed in monocyte-derived cells that constitutively express CAR infected with Ad5Luc1-PK, as compared to monocyte-derived DCs infected with Ad5Luc1-PK.
  • To further demonstrate that xeno-modified infectivity can be independent of CAR, an Ad5 Knob Inhibition assay can be performed. Monocyte-derived DCs that constitutively overexpress CAR can be infected with Ad5Luc or Ad5Luc1-PK where increasing amounts from 0-100 µg/mL of Ad5 Knob can be added to the infection mixture. The luciferase expression levels can be shown as relative light units (RLU). After infection of Ad5Luc into monocyte-derived DCs overexpressing CAR, there can be a 3-fold decrease in luciferase transgene activity when comparing infection with no Ad5 Knob present and infection with 10 µg/mL of Ad5 Knob present. Infection with 50 µg/mL or 100 µg/mL of Ad5 Knob present can further decrease the luciferase transgene activity. After infection of Ad5Luc1-PK into monocyte-derived DCs overexpressing CAR, there may be no effect on luciferase transgene activity when comparing infection with no Ad5 Knob present and infection with 10 µg/mL of Ad5 Knob present. Infection with increasing amounts of Ad5 Knob, such as 50 µg/mL or 100 µg/mL, may not affect luciferase transgene activity in DCs overexpressing CAR infected with Ad5Luc1-PK.
  • Example 10
  • This example demonstrates that the porcine knob can bind to lactose and N-acetyl-lactosamine units.
  • The affinity of porcine adenovirus knob domain for Lacto-N-neotetraose, 3-aminopropyl-lacto-N-neotetraose, 2-azidoethyl-di(N-acetyl-lactosamine), and 2-aminoethyl-tri(N-acetyl-lactosamine) can be assessed by surface plasmon resonance experiments. The galectin can be bound to a sensor chip, and the oligosaccharides can be injected at various concentrations to monitor their binding to the galectin domain. Sensorgrams can be evaluated via steady-state analysis to yield the corresponding isotherms from which the dissociation constants can be calculated. Surface plasmon resonance response can be calculated in µRIU. Lacto-N-neotetraose binds weakly to the galectin domain with a dissociation constant of 193±9µM; while 3-aminopropyl-lacto-N-neotetraose, 2-azidoethyl-di(N-acetyl-lactosamine), 2-aminoethyl-tri(N-acetyl-lactosamine) bind stronger to the galectin domain with dissociation constants of 303±4µM, 309±9µM, and 308±40µM, respectively.
  • SEQUENCE LISTING
    • <110> Washington University Curiel, David T
    • <120> PORCINE KNOB XENOTYPE CHIMERIC ADENOVIRAL VECTOR FOR DENDRITIC CELL INFECTION
    • <130> 1001-0560
    • <150> US 61/576,116
      <151> 2011-12-15
    • <160> 12
    • <170> PatentIn version 3.5
    • <210> 1
      <211> 9
      <212> PRT
      <213> Human herpesvirus 5
    • <400> 1
      Figure imgb0001
    • <210> 2
      <211> 9
      <212> PRT
      <213> Human herpesvirus 4
    • <400> 2
      Figure imgb0002
    • <210> 3
      <211> 9
      <212> PRT
      <213> Influenza A virus
    • <400> 3
      Figure imgb0003
    • <210> 4
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 4
      Figure imgb0004
    • <210> 5
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 5
      Figure imgb0005
    • <210> 6
      <211> 18
      <212> DNA
      <213> Porcine adenovirus 4
    • <400> 6
      tgtggacggg gcctgctc   18
    • <210> 7
      <211> 19
      <212> DNA
      <213> Porcine adenovirus 4
    • <400> 7
      tttattacag tatctgagg   19
    • <210> 8
      <211> 20
      <212> DNA
      <213> Human adenovirus type 5
    • <400> 8
      cagctccatc tcctaactgt   20
    • <210> 9
      <211> 20
      <212> DNA
      <213> Human adenovirus type 5
    • <400> 9
      ttcttgggca atgtatgaaa   20
    • <210> 10
      <211> 18
      <212> DNA
      <213> Porcine adenovirus 4
    • <400> 10
      tgtggacggg gcctgctc   18
    • <210> 11
      <211> 19
      <212> DNA
      <213> Porcine adenovirus 4
    • <400> 11
      tttattacag tatctgagg   19
    • <210> 12
      <211> 4
      <212> PRT
      <213> Mastadenovirus 4
    • <400> 12
      Figure imgb0006

Claims (14)

  1. A dendritic cell comprising:
    a) chimeric adenovirus-5 (Ad5) viral genome encoding:
    1) a fiber comprising a human Ad5 tail, a human Ad5 shaft and a porcine knob of porcine adenovirus type 4 (PAdV-4); and
    2) a promoter operably linked to a heterologous sequence encoding an immunizing antigen.
  2. A dendritic cell in accordance with claim 1, wherein the immunizing antigen consists of a portion of a protein expressed by a cancer cell at a level associated with a cancerous phenotype.
  3. A dendritic cell in accordance with claim 1, wherein the immunizing antigen consists of 8, from 8 to 15, or 15 contiguous amino acids.
  4. A dendritic cell in accordance with claim 1, wherein the immunizing antigen consists of 9 contiguous amino acids.
  5. A dendritic cell in accordance with claim 1, wherein the immunizing antigen consists of a sequence selected from the group consisting of NLVPMVATV (SEQ ID NO: 1), GLCTLVAML (SEQ ID NO: 2), GILGFVFTL (SEQ ID NO: 3), IMDQVPFSV (SEQ ID NO: 4), YLEPGPVTV (SEQ ID NO: 5).
  6. An ex vivo cell culture comprising a dendritic cell of claim 1.
  7. A vaccine comprising a dendritic cell of claim 1.
  8. A vaccine in accordance with claim 7, wherein the immunizing antigen is a peptide comprising 8, from 8 to 15, or 15 contiguous amino acids;
    wherein, optionally, the immunizing antigen is a peptide comprising 9 contiguous amino acids.
  9. A vaccine in accordance with claim 7, wherein the immunizing antigen is a peptide consisting of 8, from 8 to 15, or 15 contiguous amino acids;
    wherein, optionally, the immunizing antigen is a peptide consisting of 9 contiguous amino acids.
  10. A vaccine in accordance with claim 7, wherein the immunizing antigen is a peptide comprising a sequence selected from the group consisting of NLVPMVATV (SEQ ID NO: 1), GLCTLVAML (SEQ ID NO: 2), GILGFVFTL (SEQ ID NO: 3), IMDQVPFSV (SEQ ID NO: 4), YLEPGPVTV (SEQ ID NO: 5).
  11. A vaccine in accordance with claim 7, wherein the immunizing antigen is a peptide consisting of a sequence selected from the group consisting of NLVPMVATV (SEQ ID NO: 1), GLCTLVAML (SEQ ID NO: 2), GILGFVFTL (SEQ ID NO: 3), IMDQVPFSV (SEQ ID NO: 4), YLEPGPVTV (SEQ ID NO: 5).
  12. A method of transforming dendritic cells in a cell culture, the method comprising adding to a cell culture comprising dendritic cells, the chimeric adenovirus-5 (Ad5) comprising:
    1) a fiber comprising a human Ad5 tail, a human Ad5 shaft and a porcine knob of porcine adenovirus type 4 (PAdV-4); and
    2) a promoter operably linked to a heterologous sequence encoding an immunizing antigen.
  13. A method in accordance with claim 12, wherein the porcine knob comprises a galectin domain that binds to carbohydrates comprising lactose and N-acetyl-lactosamine units.
  14. A method in accordance with claim 12, wherein the porcine knob comprises a galectin domain that binds a carbohydrate structure selected from the group consisting of Galß1-4GlcNAcß1-3Galß1-4GlcNAcß1-3Galß1-4GlcNAc [tri(Nacetyl-lactosamine)], GlcNAcα1-4Galß1-4GlcNAcß1-3Galß1-4GlcNAcß1-3Galß1-4GlcNAc,Galß1-4GlcNAcß1-3Galß1-4Glc (lacto-N-neotetraose), Galα1-4Galß1-4GlcNAcß1-3Galß1-4Glc and Galß1-4GlcNAcß1-3Galß1-3GlcNAc.
EP12858372.1A 2011-12-15 2012-12-14 Porcine knob xenotype chimeric adenoviral vector for dendritic cell infection Not-in-force EP2798069B1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161576116P 2011-12-15 2011-12-15
PCT/US2012/069884 WO2013090806A2 (en) 2011-12-15 2012-12-14 Porcine knob xenotype chimeric adenoviral vector for dendritic cell infection

Publications (3)

Publication Number Publication Date
EP2798069A2 EP2798069A2 (en) 2014-11-05
EP2798069A4 EP2798069A4 (en) 2015-11-18
EP2798069B1 true EP2798069B1 (en) 2017-03-29

Family

ID=48613374

Family Applications (1)

Application Number Title Priority Date Filing Date
EP12858372.1A Not-in-force EP2798069B1 (en) 2011-12-15 2012-12-14 Porcine knob xenotype chimeric adenoviral vector for dendritic cell infection

Country Status (3)

Country Link
US (1) US9267153B2 (en)
EP (1) EP2798069B1 (en)
WO (1) WO2013090806A2 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9267153B2 (en) * 2011-12-15 2016-02-23 Washington University Porcine knob xenotype chimeric adenoviral vector for dendritic cell infection
WO2014153204A1 (en) 2013-03-14 2014-09-25 Salk Institute For Biological Studies Oncolytic adenovirus compositions
WO2017147269A1 (en) 2016-02-23 2017-08-31 Salk Institute For Biological Studies Exogenous gene expression in therapeutic adenovirus for minimal impact on viral kinetics
AU2017222568B2 (en) 2016-02-23 2020-09-10 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
CN110062630A (en) 2016-12-12 2019-07-26 萨克生物研究学院 Cancer target synthesizes adenovirus and application thereof
WO2020033876A1 (en) * 2018-08-10 2020-02-13 Nantbio, Inc. Cellular adjuvants for viral infection

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5962311A (en) * 1994-09-08 1999-10-05 Genvec, Inc. Short-shafted adenoviral fiber and its use
US7232899B2 (en) 1996-09-25 2007-06-19 The Scripps Research Institute Adenovirus vectors, packaging cell lines, compositions, and methods for preparation and use
AU744252B2 (en) * 1998-02-06 2002-02-21 Uab Research Foundation, The Adenovirus vector containing a heterologous peptide epitope in the hi loop of the fiber knob
WO2000067576A1 (en) * 1999-05-12 2000-11-16 The Uab Research Foundation Infectivity-enhanced conditionally-replicative adenovirus and uses thereof
AU2001295193A1 (en) 2000-05-01 2001-11-12 Novartis Ag Vectors for ocular transduction and use thereof for genetic therapy
EP1233059A1 (en) 2001-02-09 2002-08-21 ARTEMIS Pharmaceuticals GmbH Influenza viruses with enhanced transcriptional and replicational capacities
AU2002250081A1 (en) * 2001-02-14 2002-08-28 Uab Research Foundation Combined transductional and transcriptional targeting system for improved gene delivery
AU2002363322A1 (en) 2001-10-26 2003-05-19 Large Scale Biology Corporation Endothelial cell derived hemotopoietic growth factor
AU2003247128A1 (en) 2002-07-10 2004-02-02 Transgene S.A. Modified adenoviral fiber with ablated to cellular receptors
US20060062764A1 (en) 2004-08-25 2006-03-23 Seshidar-Reddy Police Fiber-modified adenoviral vectors for enhanced transduction of tumor cells
US7741099B2 (en) 2004-10-13 2010-06-22 Beth Israel Deaconess Medical Center Inc. Adenoviral vectors and uses thereof
WO2006119449A2 (en) * 2005-05-04 2006-11-09 Vectorlogics, Inc. Modified adenovirus containing a stabilized antibody
JP2011501958A (en) 2007-10-29 2011-01-20 バージニア テック インテレクチュアル プロパティーズ インコーポレーテッド Porcine DC-SIGN, ICAM-3 and LSECtin and their use
CA2705014A1 (en) * 2007-11-08 2009-05-14 Nestec S.A. Use of oligosaccharides containing n-acetyllactosamine for maturation of immune responses in neonates
EP2325298B1 (en) 2008-03-04 2016-10-05 The Trustees Of The University Of Pennsylvania SIMIAN ADENOVIRUSES SAdV-36, -42.1, -42.2, AND -44 AND USES THEREOF
EP2342321B1 (en) 2008-09-17 2018-04-11 Isogenis, Inc. Construction of fully-deleted adenovirus-based gene delivery vectors and uses thereof
US20100083391A1 (en) * 2008-09-26 2010-04-01 Hamilton Thomas C Cancer Compositions, Animal Models, and Methods of Use Thereof
CA2755192C (en) 2009-03-20 2018-09-11 Sangamo Biosciences, Inc. Modification of cxcr4 using engineered zinc finger proteins
US8956828B2 (en) 2009-11-10 2015-02-17 Sangamo Biosciences, Inc. Targeted disruption of T cell receptor genes using engineered zinc finger protein nucleases
US9267153B2 (en) * 2011-12-15 2016-02-23 Washington University Porcine knob xenotype chimeric adenoviral vector for dendritic cell infection

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
EP2798069A2 (en) 2014-11-05
WO2013090806A3 (en) 2014-10-02
WO2013090806A2 (en) 2013-06-20
EP2798069A4 (en) 2015-11-18
US9267153B2 (en) 2016-02-23
US20150017195A1 (en) 2015-01-15

Similar Documents

Publication Publication Date Title
EP2798069B1 (en) Porcine knob xenotype chimeric adenoviral vector for dendritic cell infection
Sirena et al. The human membrane cofactor CD46 is a receptor for species B adenovirus serotype 3
KR102044051B1 (en) Batches of recombinant adenovirus with altered terminal ends
EP3256140B1 (en) Compositions for ebola virus vaccination
US20100034774A1 (en) Serotype of adenovirus and uses thereof
MXPA06013570A (en) Chimeric adenoviruses for use in cancer treatment.
JP2024038194A (en) Delivery of sialidase to cancer cells, immune cells and tumor microenvironment
US9476061B2 (en) Adenoviral vectors for transduction of vascular tissue
Corredor et al. The non-essential left end region of the fowl adenovirus 9 genome is suitable for foreign gene insertion/replacement
JP2007527215A (en) Adenovirus E1A / E1B complementation cell line
WO2012163119A1 (en) Construction and application of mutant type b human adenovirus ad11 having enhanced oncolytic power
Fouletier-Dilling et al. Novel compound enables high-level adenovirus transduction in the absence of an adenovirus-specific receptor
Hemsath et al. Ex vivo and in vivo CD46 receptor utilization by species D human adenovirus serotype 26 (HAdV26)
Di Paolo et al. Fiber shaft-chimeric adenovirus vectors lacking the KKTK motif efficiently infect liver cells in vivo
US20100233125A1 (en) Chimeric adenovirus, method for producing the same and pharmaceutical using the same
CN115397448A (en) Delivery of sialidases to cancer cells, immune cells and tumor microenvironment
Perreau et al. The conundrum between immunological memory to adenovirus and their use as vectors in clinical gene therapy
van de Ven et al. Selective transduction of mature DC in human skin and lymph nodes by CD80/CD86-targeted fiber-modified adenovirus-5/3
Fernandes et al. Upstream bioprocess for adenovirus vectors
JP2003504316A (en) Adenovirus vectors for treating diseases
JP2011505837A (en) Vaccine against adenovirus serotype 14
WO2023047348A1 (en) Stabilized corona virus spike protein fusion proteins
Maguire Analysis of targeting approaches intended to enhance the immunogenicity of adenovirally-vectored vaccine antigens
Mercier et al. 111. A Chimeric Adenovirus Vector Encoding Reovirus Attachment Protein Sigma 1 Targets Cells Expressing Junctional Adhesion Molecule 1 and Sialic Acid
Strack Transcriptional Targeting of Dendritic Cells as a New Therapeutic Vaccine against HIV-1

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20140714

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

R17P Request for examination filed (corrected)

Effective date: 20150330

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: C12N 5/0784 20100101ALI20151001BHEP

Ipc: C12N 15/861 20060101AFI20151001BHEP

Ipc: C12N 15/63 20060101ALI20151001BHEP

Ipc: A61K 39/00 20060101ALI20151001BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20151016

17Q First examination report despatched

Effective date: 20160531

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20161028

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 879806

Country of ref document: AT

Kind code of ref document: T

Effective date: 20170415

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

RAP2 Party data changed (patent owner data changed or rights of a patent transferred)

Owner name: WASHINGTON UNIVERSITY

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602012030603

Country of ref document: DE

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602012030603

Country of ref document: DE

Representative=s name: STOLMAR & PARTNER PATENTANWAELTE PARTG MBB, DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: RENTSCH PARTNER AG, CH

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170630

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170629

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20170329

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 879806

Country of ref document: AT

Kind code of ref document: T

Effective date: 20170329

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170629

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

REG Reference to a national code

Ref country code: CH

Ref legal event code: PCAR

Free format text: NEW ADDRESS: BELLERIVESTRASSE 203 POSTFACH, 8034 ZUERICH (CH)

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170731

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170729

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 6

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602012030603

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MC

Payment date: 20171214

Year of fee payment: 6

Ref country code: LU

Payment date: 20171219

Year of fee payment: 6

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20180103

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171214

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20171231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171231

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IE

Payment date: 20181220

Year of fee payment: 7

Ref country code: DE

Payment date: 20181210

Year of fee payment: 7

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20181218

Year of fee payment: 7

Ref country code: FR

Payment date: 20181221

Year of fee payment: 7

Ref country code: CH

Payment date: 20181218

Year of fee payment: 7

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20121214

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190102

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181214

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602012030603

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170329

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20191214

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191231

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191214

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200701

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191214

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191231

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191231